Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbiology (Reading) ; 169(1)2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36748627

RESUMO

DprE2 is an essential enzyme in the synthesis of decaprenylphosphoryl-ß-d-arabinofuranose (DPA) and subsequently arabinogalactan, and is a significant new drug target for M. tuberculosis. Two compounds from the GSK-177 box set, GSK301A and GSK032A, were identified through Mt-DprE2-target overexpression studies. The Mt-DprE1-DprE2 complex was co-purified and a new in vitro DprE2 assay developed, based on the oxidation of the reduced nicotinamide adenine dinucleotide cofactor of DprE2 (NADH/NADPH). The Mt-DprE1-DprE2 complex showed interesting kinetics in both the DprE1 resazurin-based assay, where Mt-DprE2 was found to enhance Mt-DprE1 activity and reduce substrate inhibition; and also in the DprE2 assay, which similarly exhibited substrate inhibition and a difference in kinetics of the two potential cofactors, NADH and NADPH. Although, no inhibition was observed in the DprE2 assay by the two GSK set compounds, spontaneous mutant generation indicated a possible explanation in the form of a pro-drug activation pathway, involving fgd1 and fbiC.


Assuntos
Mycobacterium tuberculosis , Oxirredutases/genética , Oxirredutases/metabolismo , NAD/metabolismo , NADP/metabolismo , Antituberculosos/farmacologia , Antituberculosos/metabolismo , Proteínas de Bactérias/química
2.
Elife ; 82019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31436532

RESUMO

The immunoreceptor tyrosine-based inhibition motif (ITIM)-containing receptor G6b-B is critical for platelet production and activation. Loss of G6b-B results in severe macrothrombocytopenia, myelofibrosis and aberrant platelet function in mice and humans. Using a combination of immunohistochemistry, affinity chromatography and proteomics, we identified the extracellular matrix heparan sulfate (HS) proteoglycan perlecan as a G6b-B binding partner. Subsequent in vitro biochemical studies and a cell-based genetic screen demonstrated that the interaction is specifically mediated by the HS chains of perlecan. Biophysical analysis revealed that heparin forms a high-affinity complex with G6b-B and mediates dimerization. Using platelets from humans and genetically modified mice, we demonstrate that binding of G6b-B to HS and multivalent heparin inhibits platelet and megakaryocyte function by inducing downstream signaling via the tyrosine phosphatases Shp1 and Shp2. Our findings provide novel insights into how G6b-B is regulated and contribute to our understanding of the interaction of megakaryocytes and platelets with glycans.


Assuntos
Plaquetas/fisiologia , Heparitina Sulfato/metabolismo , Megacariócitos/fisiologia , Receptores Imunológicos/metabolismo , Animais , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Multimerização Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Transdução de Sinais
3.
J Biol Chem ; 294(18): 7348-7359, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30877199

RESUMO

A growing body of evidence implicates the mycobacterial capsule, the outermost layer of the mycobacterial cell envelope, in modulation of the host immune response and virulence of mycobacteria. Mycobacteria synthesize the dominant capsule component, α(1→4)-linked glucan, via three interconnected and potentially redundant metabolic pathways. Here, we report the crystal structure of the Mycobacterium smegmatis TreS:Pep2 complex, containing trehalose synthase (TreS) and maltokinase (Pep2), which converts trehalose to maltose 1-phosphate as part of the TreS:Pep2-GlgE pathway. The structure, at 3.6 Å resolution, revealed that a diamond-shaped TreS tetramer forms the core of the complex and that pairs of Pep2 monomers bind to opposite apices of the tetramer in a 4 + 4 configuration. However, for the M. smegmatis orthologues, results from isothermal titration calorimetry and analytical ultracentrifugation experiments indicated that the prevalent stoichiometry in solution is 4 TreS + 2 Pep2 protomers. The observed discrepancy between the crystallized complex and the behavior in the solution state may be explained by the relatively weak affinity of Pep2 for TreS (Kd 3.5 µm at mildly acidic pH) and crystal packing favoring the 4 + 4 complex. Proximity of the ATP-binding site in Pep2 to the complex interface provides a rational basis for rate enhancement of Pep2 upon binding to TreS, but the complex structure appears to rule out substrate channeling between the active sites of TreS and Pep2. Our findings provide a structural model for the trehalose synthase:maltokinase complex in M. smegmatis that offers critical insights into capsule assembly.


Assuntos
Proteínas de Bactérias/metabolismo , Glucanos/biossíntese , Glucosiltransferases/metabolismo , Mycobacterium smegmatis/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Glucosiltransferases/química , Mycobacterium smegmatis/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/química , Conformação Proteica , Soluções
4.
Acta Crystallogr D Struct Biol ; 75(Pt 1): 101-108, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30644849

RESUMO

The intracellular pathogen Mycobacterium tuberculosis is the causative agent of tuberculosis, which is a leading cause of mortality worldwide. The survival of M. tuberculosis in host macrophages through long-lasting periods of persistence depends, in part, on breaking down host cell lipids as a carbon source. The critical role of fatty-acid catabolism in this organism is underscored by the extensive redundancy of the genes implicated in ß-oxidation (∼100 genes). In a previous study, the enzymology of the M. tuberculosis L-3-hydroxyacyl-CoA dehydrogenase FadB2 was characterized. Here, the crystal structure of this enzyme in a ligand-free form is reported at 2.1 Šresolution. FadB2 crystallized as a dimer with three unique dimer copies per asymmetric unit. The structure of the monomer reveals a dual Rossmann-fold motif in the N-terminal domain, while the helical C-terminal domain mediates dimer formation. Comparison with the CoA- and NAD+-bound human orthologue mitochondrial hydroxyacyl-CoA dehydrogenase shows extensive conservation of the residues that mediate substrate and cofactor binding. Superposition with the multi-catalytic homologue M. tuberculosis FadB, which forms a trifunctional complex with the thiolase FadA, indicates that FadB has developed structural features that prevent its self-association as a dimer. Conversely, FadB2 is unable to substitute for FadB in the tetrameric FadA-FadB complex as it lacks the N-terminal hydratase domain of FadB. Instead, FadB2 may functionally (or physically) associate with the enoyl-CoA hydratase EchA8 and the thiolases FadA2, FadA3, FadA4 or FadA6 as suggested by interrogation of the STRING protein-network database.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/química , Mycobacterium tuberculosis/enzimologia , Cristalografia por Raios X , Enoil-CoA Hidratase/metabolismo , Humanos , Oxirredução , Ligação Proteica , Multimerização Proteica
5.
Sci Rep ; 8(1): 13473, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30194385

RESUMO

Nitro-substituted 1,3-benzothiazinones (nitro-BTZs) are mechanism-based covalent inhibitors of Mycobacterium tuberculosis decaprenylphosphoryl-ß-D-ribose-2'-oxidase (DprE1) with strong antimycobacterial properties. We prepared a number of oxidized and reduced forms of nitro-BTZs to probe the mechanism of inactivation of the enzyme and to identify opportunities for further chemistry. The kinetics of inactivation of DprE1 was examined using an enzymatic assay that monitored reaction progress up to 100 min, permitting compound ranking according to kinact/Ki values. The side-chain at the 2-position and heteroatom identity at the 1-position of the BTZs were found to be important for inhibitory activity. We obtained crystal structures with several compounds covalently bound. The data suggest that steps upstream from the covalent end-points are likely the key determinants of potency and reactivity. The results of protein mass spectrometry using a 7-chloro-nitro-BTZ suggest that nucleophilic reactions at the 7-position do not operate and support a previously proposed mechanism in which BTZ activation by a reduced flavin intermediate is required. Unexpectedly, a hydroxylamino-BTZ showed time-dependent inhibition and mass spectrometry corroborated that this hydroxylamino-BTZ is a mechanism-based suicide inhibitor of DprE1. With this BTZ derivative, we propose a new covalent mechanism of inhibition of DprE1 that takes advantage of the oxidation cycle of the enzyme.


Assuntos
Oxirredutases do Álcool , Antituberculosos/química , Proteínas de Bactérias , Inibidores Enzimáticos/química , Mycobacterium tuberculosis/enzimologia , Oxirredutases do Álcool/antagonistas & inibidores , Oxirredutases do Álcool/química , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Cristalografia por Raios X , Espectrometria de Massas
6.
Angew Chem Int Ed Engl ; 56(42): 13011-13015, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28815830

RESUMO

Mycobacterium tuberculosis (Mtb) DprE1, an essential isomerase for the biosynthesis of the mycobacterial cell wall, is a validated target for tuberculosis (TB) drug development. Here we report the X-ray crystal structures of DprE1 and the DprE1 resistant mutant (Y314C) in complexes with TCA1 derivatives to elucidate the molecular basis of their inhibitory activities and an unconventional resistance mechanism, which enabled us to optimize the potency of the analogs. The selected lead compound showed excellent in vitro and in vivo activities, and low risk of toxicity profile except for the inhibition of CYP2C9. A crystal structure of CYP2C9 in complex with a TCA1 analog revealed the similar interaction patterns to the DprE1-TCA1 complex. Guided by the structures, an optimized molecule was generated with differential inhibitory activities against DprE1 and CYP2C9, which provides insights for development of a clinical candidate to treat TB.


Assuntos
Antituberculosos/química , Proteínas de Bactérias/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Mycobacterium tuberculosis/metabolismo , Tiofenos/química , Animais , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Proteínas de Bactérias/antagonistas & inibidores , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Citocromo P-450 CYP2C9/química , Farmacorresistência Bacteriana/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Relação Estrutura-Atividade , Tiofenos/farmacologia , Tiofenos/uso terapêutico , Tuberculose/tratamento farmacológico , Tuberculose/veterinária
7.
Sci Rep ; 7(1): 9430, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28842600

RESUMO

Drug discovery efforts against the pathogen Mycobacterium tuberculosis (Mtb) have been advanced through phenotypic screens of extensive compound libraries. Such a screen revealed sulfolane 1 and indoline-5-sulfonamides 2 and 3 as potent inhibitors of mycobacterial growth. Optimization in the sulfolane series led to compound 4, which has proven activity in an in vivo murine model of Mtb infection. Here we identify the target and mode of inhibition of these compounds based on whole genome sequencing of spontaneous resistant mutants, which identified mutations locating to the essential α- and ß-subunits of tryptophan synthase. Over-expression studies confirmed tryptophan synthase as the biological target. Biochemical techniques probed the mechanism of inhibition, revealing the mutant enzyme complex incurs a fitness cost but does not prevent inhibitor binding. Mapping of the resistance conferring mutations onto a low-resolution crystal structure of Mtb tryptophan synthase showed they locate to the interface between the α- and ß-subunits. The discovery of anti-tubercular agents inhibiting tryptophan synthase highlights the therapeutic potential of this enzyme and draws attention to the prospect of other amino acid biosynthetic pathways as future Mtb drug targets.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Mycobacterium/efeitos dos fármacos , Mycobacterium/enzimologia , Triptofano Sintase/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Mutação , Mycobacterium/genética , Conformação Proteica , Relação Estrutura-Atividade , Tiofenos/farmacologia , Triptofano Sintase/química , Triptofano Sintase/metabolismo
8.
ACS Appl Mater Interfaces ; 9(27): 23195-23201, 2017 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-28622722

RESUMO

Supercapacitors are energy storage devices with higher energy densities than conventional capacitors but lower than batteries or fuel cells. There is a strong interest in increasing the volumetric and gravimetric capacitance of these devices to meet the growing demands of the electrical and electronic sectors. The capacitance depends largely on the electrode material, and carbon nanofibers (CNFs) have attracted much attention because of their relatively low cost, large surface area, and good electrical conductivity as well as chemical and thermal stability. The deposition of metal nanoparticles on CNFs is a promising way to increase their surface properties and, ultimately, the capacitance of the devices. In this study, nickel and silver nanoparticles were deposited on CNFs using the active screen plasma technology. The CNFs were characterized, and their electrochemical performance was assessed in a three-electrode cell. The results show significant improvements over the untreated CNFs, particularly after functionalization with silver nanoparticles.

9.
Nat Microbiol ; 1: 15006, 2016 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-27571973

RESUMO

Phenotypic screens for bactericidal compounds against drug-resistant tuberculosis are beginning to yield novel inhibitors. However, reliable target identification remains challenging. Here, we show that tetrahydropyrazo[1,5-a]pyrimidine-3-carboxamide (THPP) selectively pulls down EchA6 in a stereospecific manner, instead of the previously assigned target Mycobacterium tuberculosis MmpL3. While homologous to mammalian enoyl-coenzyme A (CoA) hydratases, EchA6 is non-catalytic yet essential and binds long-chain acyl-CoAs. THPP inhibitors compete with CoA-binding, suppress mycolic acid synthesis, and are bactericidal in a mouse model of chronic tuberculosis infection. A point mutation, W133A, abrogated THPP-binding and increased both the in vitro minimum inhibitory concentration and the in vivo effective dose 99 in mice. Surprisingly, EchA6 interacts with selected enzymes of fatty acid synthase II (FAS-II) in bacterial two-hybrid assays, suggesting essentiality may be linked to feeding long-chain fatty acids to FAS-II. Finally, our data show that spontaneous resistance-conferring mutations can potentially obscure the actual target or alternative targets of small molecule inhibitors.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Ácidos Graxos Essenciais/metabolismo , Genes Essenciais , Mycobacterium tuberculosis/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Proteínas de Ligação a Ácido Graxo/genética , Camundongos , Testes de Sensibilidade Microbiana , Mutação de Sentido Incorreto , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Mutação Puntual , Ligação Proteica , Mapeamento de Interação de Proteínas , Tuberculose/microbiologia , Tuberculose/patologia , Técnicas do Sistema de Duplo-Híbrido
10.
Open Biol ; 6(6)2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27335320

RESUMO

Mycobacterium tuberculosis (Mtb), the aetiological agent of tuberculosis, has evolved to scavenge nutrients from the confined environment of host macrophages with mycobacterial ATP-binding cassette (ABC) transporters playing a key role in nutrient acquisition. Mtb-UspC (Rv2318) is the solute-binding protein of the essential transporter UspABC, one of four Mtb ABC transporters implicated by homology in sugar acquisition. Herein, we report the structural and functional characterization of Mtb-UspC. The 1.5 Å resolution structure of UspC reveals a two subdomain architecture that forms a highly acidic carbohydrate-substrate binding cleft. This has allowed a distinct preference of Mtb-UspC for amino sugars as determined by thermal shift analysis and solution saturation transfer difference-NMR. Taken together our data support the functional assignment of UspABC as an amino-sugar transporter. Given the limited availability of carbohydrates within the phagosomal environmental niche during Mtb intracellular infection, our studies suggest that UspABC enables Mtb to optimize the use of scarce nutrients during intracellular infection, linking essentiality of this protein to a potential role in recycling components of cell-wall peptidoglycan.


Assuntos
Amino Açúcares/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Mycobacterium tuberculosis/química , Ligação Proteica , Estrutura Secundária de Proteína
11.
PLoS One ; 9(11): e113568, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25409504

RESUMO

The human pathogen Mycobacterium tuberculosis is the causative agent of pulmonary tuberculosis (TB), a disease with high worldwide mortality rates. Current treatment programs are under significant threat from multi-drug and extensively-drug resistant strains of M. tuberculosis, and it is essential to identify new inhibitors and their targets. We generated spontaneous resistant mutants in Mycobacterium bovis BCG in the presence of 10× the minimum inhibitory concentration (MIC) of compound 1, a previously identified potent inhibitor of mycobacterial growth in culture. Whole genome sequencing of two resistant mutants revealed in one case a single nucleotide polymorphism in the gene aspS at (535)GAC>(535)AAC (D179N), while in the second mutant a single nucleotide polymorphism was identified upstream of the aspS promoter region. We probed whole cell target engagement by overexpressing either M. bovis BCG aspS or Mycobacterium smegmatis aspS, which resulted in a ten-fold and greater than ten-fold increase, respectively, of the MIC against compound 1. To analyse the impact of inhibitor 1 on M. tuberculosis AspS (Mt-AspS) activity we over-expressed, purified and characterised the kinetics of this enzyme using a robust tRNA-independent assay adapted to a high-throughput screening format. Finally, to aid hit-to-lead optimization, the crystal structure of apo M. smegmatis AspS was determined to a resolution of 2.4 Å.


Assuntos
Antituberculosos/farmacologia , Aspartato-tRNA Ligase/metabolismo , Mycobacterium bovis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Piperidinas/farmacologia , Tiazóis/farmacologia , Sequência de Aminoácidos , Antituberculosos/uso terapêutico , Aspartato-tRNA Ligase/química , Aspartato-tRNA Ligase/genética , Clonagem Molecular , Cristalografia por Raios X , Dimerização , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Mycobacterium bovis/enzimologia , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/enzimologia , Mycobacterium tuberculosis/enzimologia , Piperidinas/química , Piperidinas/uso terapêutico , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Tiazóis/química , Tiazóis/uso terapêutico , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/patologia
12.
ACS Chem Biol ; 8(10): 2245-55, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23901909

RESUMO

Recent evidence established that the cell envelope of Mycobacterium tuberculosis, the bacillus causing tuberculosis (TB), is coated by an α-glucan-containing capsule that has been implicated in persistence in a mouse infection model. As one of three known metabolic routes to α-glucan in mycobacteria, the cytoplasmic GlgE-pathway converts trehalose to α(1 → 4),α(1 → 6)-linked glucan in 4 steps. Whether individual reaction steps, catalyzed by trehalose synthase TreS, maltokinase Pep2, and glycosyltransferases GlgE and GlgB, occur independently or in a coordinated fashion is not known. Here, we report the crystal structure of M. tuberculosis TreS, and show by small-angle X-ray scattering and analytical ultracentrifugation that TreS forms tetramers in solution. Together with Pep2, TreS forms a hetero-octameric complex, and we demonstrate that complex formation markedly accelerates maltokinase activity of Pep2. Thus, complex formation may act as part of a regulatory mechanism of the GlgE pathway, which overall must avoid accumulation of toxic pathway intermediates, such as maltose-1-phosphate, and optimize the use of scarce nutrients.


Assuntos
Complexos de Coordenação/metabolismo , Glucanos/biossíntese , Glucosiltransferases/metabolismo , Mycobacterium tuberculosis/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Domínio Catalítico , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Glucanos/metabolismo , Camundongos , Modelos Moleculares , Transdução de Sinais
13.
Proc Natl Acad Sci U S A ; 110(27): E2510-7, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776209

RESUMO

A cell-based phenotypic screen for inhibitors of biofilm formation in mycobacteria identified the small molecule TCA1, which has bactericidal activity against both drug-susceptible and -resistant Mycobacterium tuberculosis (Mtb) and sterilizes Mtb in vitro combined with rifampicin or isoniazid. In addition, TCA1 has bactericidal activity against nonreplicating Mtb in vitro and is efficacious in acute and chronic Mtb infection mouse models both alone and combined with rifampicin or isoniazid. Transcriptional analysis revealed that TCA1 down-regulates genes known to be involved in Mtb persistence. Genetic and affinity-based methods identified decaprenyl-phosphoryl-ß-D-ribofuranose oxidoreductase DprE1 and MoeW, enzymes involved in cell wall and molybdenum cofactor biosynthesis, respectively, as targets responsible for the activity of TCA1. These in vitro and in vivo results indicate that this compound functions by a unique mechanism and suggest that TCA1 may lead to the development of a class of antituberculosis agents.


Assuntos
Antituberculosos/farmacologia , Benzotiazóis/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Tiofenos/farmacologia , Tuberculose Pulmonar/tratamento farmacológico , Oxirredutases do Álcool , Sequência de Aminoácidos , Animais , Antituberculosos/administração & dosagem , Antituberculosos/química , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Benzotiazóis/administração & dosagem , Benzotiazóis/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Carboidratos Epimerases/antagonistas & inibidores , Carboidratos Epimerases/química , Carboidratos Epimerases/genética , Farmacorresistência Bacteriana , Feminino , Genes Bacterianos , Ensaios de Triagem em Larga Escala , Isoniazida/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , Oxirredutases/antagonistas & inibidores , Oxirredutases/química , Oxirredutases/genética , Rifampina/administração & dosagem , Tiofenos/administração & dosagem , Tiofenos/química , Tuberculose Pulmonar/microbiologia
14.
Proc Natl Acad Sci U S A ; 109(28): 11354-9, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22733761

RESUMO

Resistance against currently used antitubercular therapeutics increasingly undermines efforts to contain the worldwide tuberculosis (TB) epidemic. Recently, benzothiazinone (BTZ) inhibitors have shown nanomolar potency against both drug-susceptible and multidrug-resistant strains of the tubercle bacillus. However, their proposed mode of action is lacking structural evidence. We report here the crystal structure of the BTZ target, FAD-containing oxidoreductase Mycobacterium tuberculosis DprE1, which is essential for viability. Different crystal forms of ligand-free DprE1 reveal considerable levels of structural flexibility of two surface loops that seem to govern accessibility of the active site. Structures of complexes with the BTZ-derived nitroso derivative CT325 reveal the mode of inhibitor binding, which includes a covalent link to conserved Cys387, and reveal a trifluoromethyl group as a second key determinant of interaction with the enzyme. Surprisingly, we find that a noncovalent complex was formed between DprE1 and CT319, which is structurally identical to CT325 except for an inert nitro group replacing the reactive nitroso group. This demonstrates that binding of BTZ-class inhibitors to DprE1 is not strictly dependent on formation of the covalent link to Cys387. On the basis of the structural and activity data, we propose that the complex of DrpE1 bound to CT325 is a representative of the BTZ-target complex. These results mark a significant step forward in the characterization of a key TB drug target.


Assuntos
Benzamidas/farmacologia , Mycobacterium tuberculosis/metabolismo , Oxirredutases/química , Antibacterianos/farmacologia , Arabinose/química , Domínio Catalítico , Parede Celular/metabolismo , Chaperoninas/metabolismo , Cristalografia por Raios X/métodos , Resistência a Múltiplos Medicamentos , Inibidores Enzimáticos/farmacologia , Escherichia coli/metabolismo , Ligantes , Modelos Químicos , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Conformação Proteica
15.
PLoS Pathog ; 7(2): e1001299, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21383969

RESUMO

The D-arabinan-containing polymers arabinogalactan (AG) and lipoarabinomannan (LAM) are essential components of the unique cell envelope of the pathogen Mycobacterium tuberculosis. Biosynthesis of AG and LAM involves a series of membrane-embedded arabinofuranosyl (Araf) transferases whose structures are largely uncharacterised, despite the fact that several of them are pharmacological targets of ethambutol, a frontline drug in tuberculosis therapy. Herein, we present the crystal structure of the C-terminal hydrophilic domain of the ethambutol-sensitive Araf transferase M. tuberculosis EmbC, which is essential for LAM synthesis. The structure of the C-terminal domain of EmbC (EmbC(CT)) encompasses two sub-domains of different folds, of which subdomain II shows distinct similarity to lectin-like carbohydrate-binding modules (CBM). Co-crystallisation with a cell wall-derived di-arabinoside acceptor analogue and structural comparison with ligand-bound CBMs suggest that EmbC(CT) contains two separate carbohydrate binding sites, associated with subdomains I and II, respectively. Single-residue substitution of conserved tryptophan residues (Trp868, Trp985) at these respective sites inhibited EmbC-catalysed extension of LAM. The same substitutions differentially abrogated binding of di- and penta-arabinofuranoside acceptor analogues to EmbC(CT), linking the loss of activity to compromised acceptor substrate binding, indicating the presence of two separate carbohydrate binding sites, and demonstrating that subdomain II indeed functions as a carbohydrate-binding module. This work provides the first step towards unravelling the structure and function of a GT-C-type glycosyltransferase that is essential in M. tuberculosis.


Assuntos
Galactanos/metabolismo , Lectinas/metabolismo , Lipopolissacarídeos/metabolismo , Mycobacterium tuberculosis/enzimologia , Pentosiltransferases/química , Pentosiltransferases/metabolismo , Parede Celular/química , Parede Celular/metabolismo , Cristalografia por Raios X , Mutagênese Sítio-Dirigida , Mycobacterium smegmatis/enzimologia , Mycobacterium tuberculosis/genética , Pentosiltransferases/genética , Conformação Proteica
16.
J Bacteriol ; 192(23): 6295-301, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20870760

RESUMO

The Staphylococcus aureus autoinducer-2 (AI-2) producer protein LuxS is phosphorylated by the Ser/Thr kinase Stk1 at a unique position, Thr14. The enzymatic activity of the phosphorylated isoform of LuxS was abrogated compared to that of nonphosphorylated LuxS, thus providing the first evidence of an AI-2-producing enzyme regulated by phosphorylation and demonstrating that S. aureus possesses an original and specific system for controlling AI-2 synthesis.


Assuntos
Proteínas de Bactérias/metabolismo , Liases de Carbono-Enxofre/metabolismo , Regulação Bacteriana da Expressão Gênica , Homosserina/análogos & derivados , Proteínas Serina-Treonina Quinases/metabolismo , Staphylococcus aureus/fisiologia , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Liases de Carbono-Enxofre/genética , Homosserina/biossíntese , Lactonas , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Alinhamento de Sequência , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Treonina/metabolismo
17.
J Biol Chem ; 285(48): 37741-52, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20843801

RESUMO

Long term survival of the pathogen Mycobacterium tuberculosis in humans is linked to the immunomodulatory potential of its complex cell wall glycolipids, which include the phosphatidylinositol mannoside (PIM) series as well as the related lipomannan and lipoarabinomannan glycoconjugates. PIM biosynthesis is initiated by a set of cytosolic α-mannosyltransferases, catalyzing glycosyl transfer from the activated saccharide donor GDP-α-D-mannopyranose to the acceptor phosphatidyl-myo-inositol (PI) in an ordered and regio-specific fashion. Herein, we report the crystal structure of mannosyltransferase Corynebacterium glutamicum PimB' in complex with nucleotide to a resolution of 2.0 Å. PimB' attaches mannosyl selectively to the 6-OH of the inositol moiety of PI. Two crystal forms and GDP- versus GDP-α-d-mannopyranose-bound complexes reveal flexibility of the nucleotide conformation as well as of the structural framework of the active site. Structural comparison, docking of the saccharide acceptor, and site-directed mutagenesis pin regio-selectivity to a conserved Asp residue in the N-terminal domain that forces presentation of the correct inositol hydroxyl to the saccharide donor.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Corynebacterium glutamicum/enzimologia , Manosiltransferases/química , Manosiltransferases/genética , Fosfatidilinositóis/biossíntese , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Corynebacterium glutamicum/química , Corynebacterium glutamicum/genética , Cristalografia por Raios X , Manosiltransferases/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Especificidade por Substrato
18.
Blood ; 115(14): 2947-55, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20154219

RESUMO

The C-type lectin receptor CLEC-2 activates platelets through Src and Syk tyrosine kinases, leading to tyrosine phosphorylation of downstream adapter proteins and effector enzymes, including phospholipase-C gamma2. Signaling is initiated through phosphorylation of a single conserved tyrosine located in a YxxL sequence in the CLEC-2 cytosolic tail. The signaling pathway used by CLEC-2 shares many similarities with that used by receptors that have 1 or more copies of an immunoreceptor tyrosine-based activation motif, defined by the sequence Yxx(L/I)x(6-12)Yxx(L/I), in their cytosolic tails or associated receptor chains. Phosphorylation of the conserved immunoreceptor tyrosine-based activation motif tyrosines promotes Syk binding and activation through binding of the Syk tandem SH2 domains. In this report, we present evidence using peptide pull-down studies, surface plasmon resonance, quantitative Western blotting, tryptophan fluorescence measurements, and competition experiments that Syk activation by CLEC-2 is mediated by the cross-linking through the tandem SH2 domains with a stoichiometry of 2:1. In support of this model, cross-linking and electron microscopy demonstrate that CLEC-2 is present as a dimer in resting platelets and converted to larger complexes on activation. This is a unique mode of activation of Syk by a single YxxL-containing receptor.


Assuntos
Plaquetas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Ativação Plaquetária/fisiologia , Multimerização Proteica/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Motivos de Aminoácidos , Ativação Enzimática/fisiologia , Humanos , Fosfolipase C gama/metabolismo , Quinase Syk , Domínios de Homologia de src
19.
PLoS One ; 4(7): e6306, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19609444

RESUMO

BACKGROUND: There is an urgent need for the discovery and development of new drugs against Mycobacterium tuberculosis, the causative agent of tuberculosis, especially due to the recent emergence of multi-drug and extensively-drug resistant strains. Herein, we have examined the susceptibility of mycobacteria to the natural product platensimycin. METHODS AND FINDINGS: We have demonstrated that platensimycin has bacteriostatic activity against the fast growing Mycobacterium smegmatis (MIC = 14 microg/ml) and against Mycobacterium tuberculosis (MIC = 12 microg/ml). Growth in the presence of paltensimycin specifically inhibited the biosynthesis of mycolic acids suggesting that the antibiotic targeted the components of the mycolate biosynthesis complex. Given the inhibitory activity of platensimycin against beta-ketoacyl-ACP synthases from Staphylococcus aureus, M. tuberculosis KasA, KasB or FabH were overexpressed in M. smegmatis to establish whether these mycobacterial KAS enzymes were targets of platensimycin. In M. smegmatis overexpression of kasA or kasB increased the MIC of the strains from 14 microg/ml, to 30 and 124 microg/ml respectively. However, overexpression of fabH on did not affect the MIC. Additionally, consistent with the overexpression data, in vitro assays using purified proteins demonstrated that platensimycin inhibited Mt-KasA and Mt-KasB, but not Mt-FabH. SIGNIFICANCE: Our results have shown that platensimycin is active against mycobacterial KasA and KasB and is thus an exciting lead compound against M. tuberculosis and the development of new synthetic analogues.


Assuntos
3-Oxoacil-(Proteína de Transporte de Acila) Sintase/antagonistas & inibidores , Adamantano/farmacologia , Aminobenzoatos/farmacologia , Anilidas/farmacologia , Inibidores Enzimáticos/farmacologia , Isoenzimas/antagonistas & inibidores , Mycobacterium smegmatis/enzimologia , Mycobacterium tuberculosis/enzimologia , Sequência de Bases , Primers do DNA , Ácidos Graxos/antagonistas & inibidores , Ácidos Graxos/biossíntese , Ácidos Micólicos/antagonistas & inibidores , Ácidos Micólicos/metabolismo
20.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 4): 383-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19307721

RESUMO

The meso (or D,L) isomer of diaminopimelic acid (DAP), a precursor of L-lysine, is a key component of the pentapeptide linker in bacterial peptidoglycan. While the peptidoglycan incorporated in the highly complex cell wall of the pathogen Mycobacterium tuberculosis structurally resembles that of Escherichia coli, it is unique in that it can contain penicillin-resistant meso-DAP-->meso-DAP linkages. The interconversion of L,L-DAP and meso-DAP is catalysed by the DAP epimerase DapF, a gene product that is essential in M. tuberculosis. Here, the crystal structure of the ligand-free form of M. tuberculosis DapF (MtDapF) refined to a resolution of 2.6 A is reported. MtDapF shows small if distinct deviations in secondary structure from the two-domain alpha/beta-fold of the known structures of Haemophilus influenzae DapF and Bacillus anthracis DapF, which are in line with its low sequence identity (

Assuntos
Isomerases de Aminoácido/química , Proteínas de Bactérias/química , Mycobacterium tuberculosis/enzimologia , Bacillus anthracis/enzimologia , Domínio Catalítico , Cristalografia por Raios X , Haemophilus influenzae/enzimologia , Modelos Químicos , Modelos Moleculares , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...